Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Neurosci ; 43(49): 8336-8347, 2023 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-37845032

RESUMEN

Many glutamatergic synapse proteins contain a 4.1N protein binding domain. However, a role for 4.1N in the regulation of glutamatergic neurotransmission has been controversial. Here, we observe significantly higher expression of protein 4.1N in granule neurons of the dentate gyrus (DG granule neurons) compared with other hippocampal regions. We discover that reducing 4.1N expression in rat DG granule neurons of either sex results in a significant reduction in glutamatergic synapse function that is caused by a decrease in the number of glutamatergic synapses. By contrast, we find reduction of 4.1N expression in hippocampal CA1 pyramidal neurons has no impact on basal glutamatergic neurotransmission. We also find 4.1N's C-terminal domain (CTD) to be nonessential to its role in the regulation of glutamatergic synapses of DG granule neurons. Instead, we show that 4.1N's four-point-one, ezrin, radixin, and moesin (FERM) domain is essential for supporting synaptic AMPA receptor (AMPAR) function in these neurons. Altogether, this work demonstrates a novel, cell type-specific role for protein 4.1N in governing glutamatergic synapse function.SIGNIFICANCE STATEMENT Glutamatergic synapses exhibit immense molecular diversity. In comparison to heavily studied Schaffer collateral, CA1 glutamatergic synapses, significantly less is known about perforant path-dentate gyrus (DG) synapses. Our data demonstrate that compromising 4.1N function in CA1 pyramidal neurons produces no alteration in basal glutamatergic synaptic transmission. However, in DG granule neurons, compromising 4.1N function leads to a significant decrease in the strength of glutamatergic neurotransmission at perforant pathway synapses. Together, our data identifies 4.1N as a cell type-specific regulator of synaptic transmission within the hippocampus and reveals a unique molecular program that governs perforant pathway synapse function.


Asunto(s)
Hipocampo , Sinapsis , Ratas , Animales , Hipocampo/fisiología , Sinapsis/fisiología , Transmisión Sináptica , Neuronas/fisiología , Vía Perforante/fisiología , Giro Dentado/fisiología
2.
J Neurosci ; 41(37): 7768-7778, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34353896

RESUMEN

We recently identified an autism spectrum disorder/intellectual disability (ASD/ID)-related de novo mutation hotspot in the Rac1-activating GEF1 domain of the protein Trio. Trio is a Rho guanine nucleotide exchange factor (RhoGEF) that is essential for glutamatergic synapse function. An ASD/ID-related mutation identified in Trio's GEF1 domain, Trio D1368V, produces a pathologic increase in glutamatergic synaptogenesis, suggesting that Trio is coupled to synaptic regulatory mechanisms that govern glutamatergic synapse formation. However, the molecular mechanisms by which Trio regulates glutamatergic synapses are largely unexplored. Here, using biochemical methods, we identify an interaction between Trio and the synaptogenic protein Neuroligin 1 (NLGN1) in the brain. Molecular biological approaches were then combined with super-resolution dendritic spine imaging and whole-cell voltage-clamp electrophysiology in hippocampal slices from male and female rats to examine the impact ASD/ID-related Trio mutations have on NLGN1-mediated synaptogenesis. We find that an ASD/ID-related mutation in Trio's eighth spectrin repeat region, Trio N1080I, inhibits Trio's interaction with NLGN1 and prevents Trio D1368V-mediated synaptogenesis. Inhibiting Trio's interaction with NLGN1 via Trio N1080I blocked NLGN1-mediated synaptogenesis and increases in synaptic NMDA Receptor function but not NLGN1-mediated increases in synaptic AMPA Receptor function. Finally, we show that the aberrant synaptogenesis produced by Trio D1368V is dependent on NLGN signaling. Our findings demonstrate that ASD/ID-related mutations in Trio are able to pathologically increase as well as decrease NLGN-mediated effects on glutamatergic neurotransmission, and point to an NLGN1-Trio interaction as part of a key pathway involved in ASD/ID etiology.SIGNIFICANCE STATEMENT A number of genes have been implicated in the development of autism spectrum disorder/intellectual disability (ASD/ID) in humans. It is now important to identify relationships between these genes to uncover specific cellular regulatory pathways that contribute to these disorders. In this study, we discover that two glutamatergic synapse regulatory proteins implicated in ASD/ID, Trio and Neuroligin 1, interact with one another to promote glutamatergic synaptogenesis. We also identify ASD/ID-related mutations in Trio that either inhibit or augment Neuroligin 1-mediated glutamatergic synapse formation. Together, our results identify a synaptic regulatory pathway that, when disrupted, likely contributes to the development of ASD/ID. Going forward, it will be important to determine whether this pathway represents a point of convergence of other proteins implicated in ASD/ID.


Asunto(s)
Trastorno del Espectro Autista/genética , Moléculas de Adhesión Celular Neuronal/genética , Discapacidad Intelectual/genética , Mutación , Sinapsis/genética , Animales , Trastorno del Espectro Autista/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Hipocampo/metabolismo , Discapacidad Intelectual/metabolismo , Masculino , Neurogénesis/fisiología , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo
3.
J Neurosci ; 39(47): 9306-9315, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31597723

RESUMEN

Mounting evidence suggests numerous glutamatergic synapse subtypes exist in the brain, and that these subtypes are likely defined by unique molecular regulatory mechanisms. Recent work has identified substantial divergence of molecular composition between commonly studied Schaffer collateral synapses and perforant path-dentate gyrus (DG) synapses of the hippocampus. However, little is known about the molecular mechanisms that may confer unique properties to perforant path-DG synapses. Here we investigate whether the RhoGEF (Rho guanine-nucleotide exchange factor) protein Tiam1 plays a unique role in the regulation of glutamatergic synapses in dentate granule neurons using a combination of molecular, electrophysiological, and imaging approaches in rat entorhino-hippocampal slices of both sexes. We find that inhibition of Tiam1 function in dentate granule neurons reduces synaptic AMPA receptor function and causes dendritic spines to adopt an elongated filopodia-like morphology. We also find that Tiam1's support of perforant path-DG synapse function is dependent on its GEF domain and identify a potential role for the auto-inhibitory PH domain of Tiam1 in regulating Tiam1 function at these synapses. In marked contrast, reduced Tiam1 expression in CA1 pyramidal neurons produced no effect on glutamatergic synapse development. Together, these data identify a critical role for Tiam1 in the hippocampus and reveal a unique Tiam1-mediated molecular program of glutamatergic synapse regulation in dentate granule neurons.SIGNIFICANCE STATEMENT Several lines of evidence independently point to the molecular diversity of glutamatergic synapses in the brain. Rho guanine-nucleotide exchange factor (RhoGEF) proteins as powerful modulators of glutamatergic synapse function have also become increasingly appreciated in recent years. Here we investigate the synaptic regulatory role of the RhoGEF protein Tiam1, whose expression appears to be remarkably enriched in granule neurons of the dentate gyrus. We find that Tiam1 plays a critical role in the development of glutamatergic perforant path-dentate gyrus synapses, but not in commonly studied in Schaffer collateral-CA1 synapses. Together, these data reveal a unique RhoGEF-mediated molecular program of glutamatergic synapse regulation in dentate granule neurons.


Asunto(s)
Ácido Glutámico/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Sinapsis/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/fisiología , Animales , Animales Recién Nacidos , Giro Dentado/química , Giro Dentado/citología , Giro Dentado/metabolismo , Femenino , Ácido Glutámico/análisis , Hipocampo/química , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Sinapsis/química , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/análisis
4.
Annu Rev Physiol ; 78: 351-65, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26863325

RESUMEN

For more than 20 years, we have known that Ca(2+)/calmodulin-dependent protein kinase (CaMKII) activation is both necessary and sufficient for the induction of long-term potentiation (LTP). During this time, tremendous effort has been spent in attempting to understand how CaMKII activation gives rise to this phenomenon. Despite such efforts, there is much to be learned about the molecular mechanisms involved in LTP induction downstream of CaMKII activation. In this review, we highlight recent developments that have shaped our current thinking about the molecular mechanisms underlying LTP and discuss important questions that remain in the field.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Potenciación a Largo Plazo/fisiología , Transporte de Proteínas/fisiología , Receptores AMPA/metabolismo , Animales , Humanos
5.
Proc Natl Acad Sci U S A ; 113(8): 2264-9, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858404

RESUMEN

The molecular mechanism underlying long-term potentiation (LTP) is critical for understanding learning and memory. CaMKII, a key kinase involved in LTP, is both necessary and sufficient for LTP induction. However, how CaMKII gives rise to LTP is currently unknown. Recent studies suggest that Rho GTPases are necessary for LTP. Rho GTPases are activated by Rho guanine exchange factors (RhoGEFs), but the RhoGEF(s) required for LTP also remain unknown. Here, using a combination of molecular, electrophysiological, and imaging techniques, we show that the RhoGEF Kalirin and its paralog Trio play critical and redundant roles in excitatory synapse structure and function. Furthermore, we show that CaMKII phosphorylation of Kalirin is sufficient to enhance synaptic AMPA receptor expression, and that preventing CaMKII signaling through Kalirin and Trio prevents LTP induction. Thus, our data identify Kalirin and Trio as the elusive targets of CaMKII phosphorylation responsible for AMPA receptor up-regulation during LTP.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Potenciación a Largo Plazo/fisiología , Proteínas del Tejido Nervioso/fisiología , Transmisión Sináptica/fisiología , Animales , Región CA1 Hipocampal/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Humanos , Técnicas In Vitro , Ratones , Modelos Neurológicos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Ratas , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Regulación hacia Arriba
6.
J Neurosci ; 35(28): 10168-71, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26180193

RESUMEN

Recent evidence has resurrected the idea that the amino acid aspartate, a selective NMDA receptor agonist, is a neurotransmitter. Using a mouse that lacks the glutamate-selective vesicular transporter VGLUT1, we find that glutamate alone fully accounts for the activation of NMDA receptors at excitatory synapses in the hippocampus. This excludes a role for aspartate and, by extension, a recently proposed role for the sialic acid transporter sialin in excitatory transmission. SIGNIFICANCE STATEMENT: It has been proposed that the amino acid aspartate serves as a neurotransmitter. Although aspartate is a selective agonist for NMDA receptors, we find that glutamate alone fully accounts for neurotransmission at excitatory synapses in the hippocampus, excluding a role for aspartate.


Asunto(s)
Ácido Aspártico/metabolismo , Neuronas/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Ácido Aspártico/farmacología , Región CA1 Hipocampal/citología , Estimulación Eléctrica , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/genética , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Técnicas In Vitro , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , ARN Mensajero/metabolismo , Estadísticas no Paramétricas , Proteína 1 de Transporte Vesicular de Glutamato/deficiencia , Proteína 1 de Transporte Vesicular de Glutamato/genética
7.
Proc Natl Acad Sci U S A ; 109(47): 19426-31, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23129617

RESUMEN

AMPA receptors (AMPARs) mediate the majority of fast excitatory neurotransmission, and their density at postsynaptic sites determines synaptic strength. Ubiquitination is a posttranslational modification that dynamically regulates the synaptic expression of many proteins. However, very few of the ubiquitinating enzymes implicated in the process have been identified. In a screen to identify transmembrane RING domain-containing E3 ubiquitin ligases that regulate surface expression of AMPARs, we identified RNF167. Predominantly lysosomal, a subpopulation of RNF167 is located on the surface of cultured neurons. Using a RING mutant RNF167 or a specific shRNA to eliminate endogenous RNF167, we demonstrate that AMPAR surface expression increases in hippocampal neurons with disrupted RNF167 activity and that RNF167 is involved in activity-dependent ubiquitination of AMPARs. In addition, RNF167 regulates synaptic AMPAR currents, whereas synaptic NMDAR currents are unaffected. Therefore, our study identifies RNF167 as a selective regulator of AMPAR-mediated neurotransmission and expands our understanding of how ubiquitination dynamically regulates excitatory synapses.


Asunto(s)
Receptores AMPA/metabolismo , Transmisión Sináptica , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Hipocampo/citología , Humanos , Lisosomas/metabolismo , Neuronas/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
J Neurosci ; 33(33): 13312-9, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23946389

RESUMEN

The extensive dendritic arbor of a pyramidal cell introduces considerable complexity to the integration of synaptic potentials. Propagation of dendritic potentials is largely passive, in contrast to regenerative axonal potentials that are maintained by voltage-gated sodium channels, leading to a declination in amplitude as dendritic potentials travel toward the soma in a manner that disproportionally affects distal synaptic inputs. To counteract this amplitude filtering, Schaffer collateral synapses onto CA1 pyramidal cells contain a varying number of AMPA receptors (AMPARs) per synapse that increases with distance from the soma, a phenomenon known as distance-dependent scaling. Here, we undertake an investigation into the molecular mechanisms of distance-dependent scaling. Using dendritic recordings from rat pyramidal neurons, we confirm the basic scaling phenomenon and find that it is expressed and can be manipulated cell autonomously. Finally, we show that it depends on the presence of both a reserve pool of AMPARs and the AMPAR subunit GluA2.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Dendritas/metabolismo , Células Piramidales/metabolismo , Receptores AMPA/biosíntesis , Animales , Western Blotting , Células Cultivadas , Potenciales Postsinápticos Excitadores/fisiología , Células HEK293 , Humanos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Transmisión Sináptica/fisiología
9.
eNeuro ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871458

RESUMEN

Glutamatergic synapses exhibit significant molecular diversity but circuit-specific mechanisms that underlie synaptic regulation are not well characterized. Prior reports show that RhoGEF Tiam1 regulates perforant path-dentate gyrus (DG) granule neuron synapses. In the present study, we report Tiam1's homolog Tiam2 is implicated in glutamatergic neurotransmission at CA1 pyramidal neurons. We find that Tiam2 regulates evoked excitatory glutamatergic currents via a post-synaptic mechanism mediated by the catalytic Dbl-homology domain. Overall, we present evidence for RhoGEF Tiam2's role in glutamatergic synapse function at Schaffer-collateral-CA1 pyramidal neuron synapses.Significance statement Glutamatergic synapses are known to vary in composition and function but how this heterogeneity is established to create input-specific synaptic diversity is not well understood. In the present study we show Tiam2 regulates glutamatergic neurotransmission at Schaffer-collateral-CA1 pyramidal neuron synapses. We find that this function is dependent on its catalytic domain. By contrast we did not observe a role for Tiam2 in synaptic transmission at perforant path-DG granule neuron synapses. We also find that Tiam1 and Tiam2 are individually dispensable for functional synaptic plasticity in CA1 pyramidal neurons. To our knowledge, this is the first evidence of the RhoGEF Tiam2's role in regulating glutamatergic synapses.

10.
Mol Autism ; 15(1): 12, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38566250

RESUMEN

BACKGROUND: Glutamatergic synapse dysfunction is believed to underlie the development of Autism Spectrum Disorder (ASD) and Intellectual Disability (ID) in many individuals. However, identification of genetic markers that contribute to synaptic dysfunction in these individuals is notoriously difficult. Based on genomic analysis, structural modeling, and functional data, we recently established the involvement of the TRIO-RAC1 pathway in ASD and ID. Furthermore, we identified a pathological de novo missense mutation hotspot in TRIO's GEF1 domain. ASD/ID-related missense mutations within this domain compromise glutamatergic synapse function and likely contribute to the development of ASD/ID. The number of ASD/ID cases with mutations identified within TRIO's GEF1 domain is increasing. However, tools for accurately predicting whether such mutations are detrimental to protein function are lacking. METHODS: Here we deployed advanced protein structural modeling techniques to predict potential de novo pathogenic and benign mutations within TRIO's GEF1 domain. Mutant TRIO-9 constructs were generated and expressed in CA1 pyramidal neurons of organotypic cultured hippocampal slices. AMPA receptor-mediated postsynaptic currents were examined in these neurons using dual whole-cell patch clamp electrophysiology. We also validated these findings using orthogonal co-immunoprecipitation and fluorescence lifetime imaging (FLIM-FRET) experiments to assay TRIO mutant overexpression effects on TRIO-RAC1 binding and on RAC1 activity in HEK293/T cells. RESULTS: Missense mutations in TRIO's GEF1 domain that were predicted to disrupt TRIO-RAC1 binding or stability were tested experimentally and found to greatly impair TRIO-9's influence on glutamatergic synapse function. In contrast, missense mutations in TRIO's GEF1 domain that were predicted to have minimal effect on TRIO-RAC1 binding or stability did not impair TRIO-9's influence on glutamatergic synapse function in our experimental assays. In orthogonal assays, we find most of the mutations predicted to disrupt binding display loss of function but mutants predicted to disrupt stability do not reflect our results from neuronal electrophysiological data. LIMITATIONS: We present a method to predict missense mutations in TRIO's GEF1 domain that may compromise TRIO function and test for effects in a limited number of assays. Possible limitations arising from the model systems employed here can be addressed in future studies. Our method does not provide evidence for whether these mutations confer ASD/ID risk or the likelihood that such mutations will result in the development of ASD/ID. CONCLUSIONS: Here we show that a combination of structure-based computational predictions and experimental validation can be employed to reliably predict whether missense mutations in the human TRIO gene impede TRIO protein function and compromise TRIO's role in glutamatergic synapse regulation. With the growing accessibility of genome sequencing, the use of such tools in the accurate identification of pathological mutations will be instrumental in diagnostics of ASD/ID.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Células HEK293 , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Mutación , Mutación Missense , Neuronas/metabolismo
11.
J Neurophysiol ; 109(3): 758-67, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23136341

RESUMEN

General anesthetics produce anesthesia by depressing central nervous system activity. Activation of inhibitory GABA(A) receptors plays a central role in the action of many clinically relevant general anesthetics. Even so, there is growing evidence that anesthetics can act at a presynaptic locus to inhibit neurotransmitter release. Our own data identified the neurotransmitter release machinery as a target for anesthetic action. In the present study, we sought to examine the site of anesthetic action more closely. Exocytosis was stimulated by directly elevating the intracellular Ca(2+) concentration at neurotransmitter release sites, thereby bypassing anesthetic effects on channels and receptors, allowing anesthetic effects on the neurotransmitter release machinery to be examined in isolation. Three different PC12 cell lines, which had the expression of different release machinery proteins stably suppressed by RNA interference, were used in these studies. Interestingly, there was still significant neurotransmitter release when these knockdown PC12 cells were stimulated. We have previously shown that etomidate, isoflurane, and propofol all inhibited the neurotransmitter release machinery in wild-type PC12 cells. In the present study, we show that knocking down synaptotagmin I completely prevented etomidate from inhibiting neurotransmitter release. Synaptotagmin I knockdown also diminished the inhibition produced by propofol and isoflurane, but the magnitude of the effect was not as large. Knockdown of SNAP-25 and SNAP-23 expression also changed the ability of these three anesthetics to inhibit neurotransmitter release. Our results suggest that general anesthetics inhibit the neurotransmitter release machinery by interacting with multiple SNARE and SNARE-associated proteins.


Asunto(s)
Anestésicos Generales/farmacología , Catecolaminas/metabolismo , Exocitosis/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/genética , Sinaptotagmina I/genética , Animales , Etomidato/farmacología , Expresión Génica , Isoflurano/farmacología , Células PC12 , Propofol/farmacología , ARN Interferente Pequeño , Ratas , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sinaptotagmina I/metabolismo
12.
bioRxiv ; 2023 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-37745471

RESUMEN

Neural circuits, which constitute the substrate for brain processing, can be traced in the retrograde direction, from postsynaptic to presynaptic cells, using methods based on introducing modified rabies virus into genetically marked cell types. These methods have revolutionized the field of neuroscience. However, similarly reliable, transsynaptic, and non-toxic methods to trace circuits in the anterograde direction are not available. Here, we describe such a method based on an antibody-like protein selected against the extracellular N-terminus of the AMPA receptor subunit GluA1 (AMPA.FingR). ATLAS (Anterograde Transsynaptic Label based on Antibody-like Sensors) is engineered to release the AMPA.FingR and its payload, which can include Cre recombinase, from presynaptic sites into the synaptic cleft, after which it binds to GluA1, enters postsynaptic cells through endocytosis and subsequently carries its payload to the nucleus. Testing in vivo and in dissociated cultures shows that ATLAS mediates monosynaptic tracing from genetically determined cells that is strictly anterograde, synaptic, and non-toxic. Moreover, ATLAS shows activity dependence, which may make tracing active circuits that underlie specific behaviors possible.

13.
Nat Commun ; 13(1): 798, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35145085

RESUMEN

Mutations in the putative glutamatergic synapse scaffolding protein SAP97 are associated with the development of schizophrenia in humans. However, the role of SAP97 in synaptic regulation is unclear. Here we show that SAP97 is expressed in the dendrites of granule neurons in the dentate gyrus but not in the dendrites of other hippocampal neurons. Schizophrenia-related perturbations of SAP97 did not affect CA1 pyramidal neuron synapse function. Conversely, these perturbations produce dramatic augmentation of glutamatergic neurotransmission in granule neurons that can be attributed to a release of perisynaptic GluA1-containing AMPA receptors into the postsynaptic densities of perforant pathway synapses. Furthermore, inhibiting SAP97 function in the dentate gyrus was sufficient to impair contextual episodic memory. Together, our results identify a cell-type-specific synaptic regulatory mechanism in the dentate gyrus that, when disrupted, impairs contextual information processing in rats.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Giro Dentado/fisiología , Proteínas de la Membrana/genética , Memoria Episódica , Mutación , Esquizofrenia/genética , Sinapsis/metabolismo , Animales , Femenino , Hipocampo/metabolismo , Masculino , Neuronas/metabolismo , Densidad Postsináptica/metabolismo , Células Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Esquizofrenia/metabolismo , Transmisión Sináptica/fisiología
14.
J Physiol ; 589(Pt 5): 1103-15, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21173083

RESUMEN

The mechanism of general anaesthetic action is only partially understood. Facilitation of inhibitory GABAA receptors plays an important role in the action of most anaesthetics, but is thought to be especially relevant in the case of intravenous anaesthetics, like etomidate and propofol. Recent evidence suggests that anaesthetics also inhibit excitatory synaptic transmission via a presynaptic mechanism(s), but it has been difficult to determine whether these agents act on the neurotransmitter release machinery itself. In the present study we sought to determine whether the intravenous anaesthetics propofol and etomidate inhibit the release machinery. For these studies we used an experimental approach that directly regulated [Ca2+]i at neurotransmitter release sites, thereby bypassing anaesthetic effects on channels and receptors in order to allow anaesthetic effects on the neurotransmitter release machinery to be examined in isolation. The data show that clinically relevant concentrations of propofol and etomidate inhibited the neurotransmitter release machinery in neurosecretory cells and in cultured hippocampal neurons. md130A is a mutant form of syntaxin with a truncated C-terminus. Overexpressing md130A in PC12 cells completely eliminated the reduction in neurotransmitter release produced by propofol, without affecting release itself. In contrast, overexpressing md130A in PC12 cells had little or no effect on the response to etomidate. These results suggest that both propofol and etomidate inhibit neurotransmitter release by a direct interaction with SNAREs and/or SNARE-associated proteins but they do so at different sites.


Asunto(s)
Anestésicos Intravenosos/farmacología , Etomidato/farmacología , Neuronas/efectos de los fármacos , Propofol/farmacología , Transmisión Sináptica/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Neuronas/metabolismo , Células PC12 , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo
15.
Sci Rep ; 11(1): 11329, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34059719

RESUMEN

While efficient methods are well established for studying postsynaptic protein regulation of glutamatergic synapses in the mammalian central nervous system, similarly efficient methods are lacking for studying proteins regulating presynaptic function. In the present study, we introduce an optical/electrophysiological method for investigating presynaptic molecular regulation. Here, using an optogenetic approach, we selectively stimulate genetically modified presynaptic CA3 pyramidal neurons in the hippocampus and measure optically-induced excitatory postsynaptic currents produced in unmodified postsynaptic CA1 pyramidal neurons. While such use of optogenetics is not novel, previous implementation methods do not allow basic quantification of the changes in synaptic strength produced by genetic manipulations. We find that incorporating simultaneous recordings of fiber volley amplitude provides a control for optical stimulation intensity and, as a result, creates a metric of synaptic efficacy that can be compared across experimental conditions. In the present study, we utilize our new method to demonstrate that inhibition of synaptotagmin 1 expression in CA3 pyramidal neurons leads to a significant reduction in Schaffer collateral synapse function, an effect that is masked with conventional electrical stimulation. Our hope is that this method will expedite our understanding of molecular regulatory pathways that govern presynaptic function.


Asunto(s)
Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores , Optogenética/métodos , Terminales Presinápticos/fisiología , Animales , Ratas
16.
Trends Neurosci ; 43(7): 505-518, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32513570

RESUMEN

Changes in the actin cytoskeleton are a primary mechanism mediating the morphological and functional plasticity that underlies learning and memory. The synaptic Ras homologous (Rho) guanine nucleotide exchange factors (GEFs) Kalirin and Trio have emerged as central regulators of actin dynamics at the synapse. The increased attention surrounding Kalirin and Trio stems from the growing evidence for their roles in the etiology of a wide range of neurodevelopmental and neurodegenerative disorders. In this Review, we discuss recent findings revealing the unique and diverse functions of these paralog proteins in neurodevelopment, excitatory synaptic transmission, and plasticity. We additionally survey the growing literature implicating these proteins in various neurological disorders.


Asunto(s)
Encefalopatías , Transmisión Sináptica , Actinas , Humanos , Factores de Intercambio de Guanina Nucleótido Rho , Sinapsis
17.
Cell Rep ; 29(10): 2944-2952.e5, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31801062

RESUMEN

The RhoGEFs Kalirin-7 and Trio are regulators of synaptic plasticity, and their dysregulation is associated with a range of neurodevelopmental and neurodegenerative disorders. Although studies have implicated both Kalirin and Trio in certain diseases, such as tauopathies, they remarkably differ in their association with other disorders. Using unbiased proteomics, we identified interactomes of Kalirin-7 and Trio to ascertain distinct protein association networks associated with their respective function and revealed groups of proteins that preferentially interact with a particular RhoGEF. In comparison, we find Trio interacts with a range of axon guidance and presynaptic complexes, whereas Kalirin-7 associates with several synaptic adhesion molecules. Specifically, we show Kalirin-7 is an interactor of the cell adhesion molecule neuroligin-1 (NLGN1), and NLGN1-dependent synaptic function is mediated through Kalirin-7 in an interaction-dependent manner. Our data reveal not only the interactomes of two important disease-related proteins, but also provide an intracellular effector of NLGN1 function.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas Sprague-Dawley , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Sinapsis/metabolismo
18.
Nat Commun ; 10(1): 2612, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31197141

RESUMEN

Primary microcephaly is caused by mutations in genes encoding centrosomal proteins including WDR62 and KIF2A. However, mechanisms underlying human microcephaly remain elusive. By creating mutant mice and human cerebral organoids, here we found that WDR62 deletion resulted in a reduction in the size of mouse brains and organoids due to the disruption of neural progenitor cells (NPCs), including outer radial glia (oRG). WDR62 ablation led to retarded cilium disassembly, long cilium, and delayed cell cycle progression leading to decreased proliferation and premature differentiation of NPCs. Mechanistically, WDR62 interacts with and promotes CEP170's localization to the basal body of primary cilium, where CEP170 recruits microtubule-depolymerizing factor KIF2A to disassemble cilium. WDR62 depletion reduced KIF2A's basal body localization, and enhanced KIF2A expression partially rescued deficits in cilium length and NPC proliferation. Thus, modeling microcephaly with cerebral organoids and mice reveals a WDR62-CEP170-KIF2A pathway promoting cilium disassembly, disruption of which contributes to microcephaly.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinesinas/metabolismo , Microcefalia/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Animales , Técnicas de Cultivo de Célula , Proteínas de Ciclo Celular/genética , Diferenciación Celular , Línea Celular , Proliferación Celular , Cilios/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Noqueados , Microcefalia/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Células-Madre Neurales/patología , Neuroglía/citología , Neuroglía/patología , Organoides/patología , Fosfoproteínas/genética , ARN Interferente Pequeño/metabolismo
19.
Front Mol Neurosci ; 11: 223, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30042656

RESUMEN

The small GTPase Rac1 promotes actin polymerization and plays a critical and increasingly appreciated role in the development and plasticity of glutamatergic synapses. Growing evidence suggests that disruption of the Rac1 signaling pathway at glutamatergic synapses contributes to Autism Spectrum Disorder/intellectual disability (ASD/ID)-related behaviors seen in animal models of ASD/ID. Rac1 has also been proposed as a strong candidate of convergence for many factors implicated in the development of ASD/ID. However, the effects of ASD/ID-related mutations in Rac1 itself have not been explored in neurons. Here, we investigate a recently reported de novo missense mutation in Rac1 found in an individual with severe ID. Our modeling predicts that this mutation will strongly inhibit Rac1 activation by occluding Rac1's GTP binding pocket. Indeed, we find that this de novo mutation prevents Rac1 function and results in a selective reduction in synaptic AMPA receptor function. Furthermore, this mutation prevents the induction of long-term potentiation (LTP), the cellular mechanism underlying learning and memory formation. Together, our findings strongly suggest that this mutation contributes to the development of ID in this individual. This research demonstrates the importance of Rac1 in synaptic function and plasticity and contributes to a growing body of evidence pointing to dysregulation of actin polymerization at glutamatergic synapses as a contributing factor to ASD/ID.

20.
Nat Commun ; 8(1): 601, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928363

RESUMEN

The Rho guanine nucleotide exchange factor (RhoGEF) Trio promotes actin polymerization by directly activating the small GTPase Rac1. Recent studies suggest that autism spectrum disorder (ASD)-related behavioral phenotypes in animal models of ASD can be produced by dysregulation of Rac1's control of actin polymerization at glutamatergic synapses. Here, in humans, we discover a large cluster of ASD-related de novo mutations in Trio's Rac1 activating domain, GEF1. Our study reveals that these mutations produce either hypofunctional or hyperfunctional forms of Trio in rodent neurons in vitro. In accordance with pathological increases or decreases in glutamatergic neurotransmission observed in animal models of ASD, we find that these mutations result in either reduced synaptic AMPA receptor expression or enhanced glutamatergic synaptogenesis. Together, our findings implicate both excessive and reduced Trio activity and the resulting synaptic dysfunction in ASD-related pathogenesis, and point to the Trio-Rac1 pathway at glutamatergic synapses as a possible key point of convergence of many ASD-related genes.Trio is a RhoGEF protein that promotes actin polymerization and is implicated in the regulation of glutamatergic synapses in autism spectrum disorder (ASD). Here the authors identify a large cluster of de novo mutations in the GEF1 domain of Trio in whole-exome sequencing data from individuals with ASD, and confirm that some of these mutations lead to glutamatergic dysregulation in vitro.


Asunto(s)
Trastorno del Espectro Autista/genética , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Animales , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/fisiopatología , Femenino , Predisposición Genética a la Enfermedad , Ácido Glutámico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Técnicas In Vitro , Masculino , Mutación , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA